Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
Add more filters











Publication year range
1.
J Biol Chem ; 276(13): 10063-71, 2001 Mar 30.
Article in English | MEDLINE | ID: mdl-11139576

ABSTRACT

Recombinant nematode anticoagulant protein c2 (rNAPc2) is a potent, factor Xa (fXa)-dependent small protein inhibitor of factor VIIa-tissue factor (fVIIa.TF), which binds to a site on fXa that is distinct from the catalytic center (exo-site). In the present study, the role of other fX derivatives in presenting rNAPc2 to fVIIa.TF is investigated. Catalytically active and active site blocked fXa, as well as a plasma-derived and an activation-resistant mutant of zymogen fX bound to rNAPc2 with comparable affinities (K(D) = 1-10 nm), and similarly supported the inhibition of fVIIa.TF (K(i)* = approximately 10 pm). The roles of phospholipid membrane composition in the inhibition of fVIIa.TF by rNAPc2 were investigated using TF that was either detergent-solubilized (TF(S)), or reconstituted into membranes, containing phosphatidylcholine (TF(PC)) or a mixture of phosphatidylcholine and phosphatidylserine (TF(PCPS)). In the absence of the fX derivative, inhibition of fVIIa.TF was similar for all three conditions (K(i) approximately 1 microm), whereas the addition of the fX derivative increased the respective inhibition by 35-, 150-, or 100,000-fold for TF(S), TF(PC), and TF(PCPS). The removal of the gamma-carboxyglutamic acid-containing domain from the fX derivative did not affect the binding to rNAPc2, but abolished the effect of factor Xa as a scaffold for the inhibition of fVIIa.TF by rNAPc2. The overall anticoagulant potency of rNAPc2, therefore, results from a coordinated recognition of an exo-site on fX/fXa and of the active site of fVIIa, both of which are properly positioned in the ternary fVIIa.TF.fX(a) complex assembled on an appropriate phospholipid surface.


Subject(s)
Cysteine Endopeptidases/chemistry , Enzyme Precursors/metabolism , Factor VIIa/metabolism , Factor Xa/metabolism , Helminth Proteins/metabolism , Neoplasm Proteins , Recombinant Proteins/metabolism , Animals , Anticoagulants/pharmacology , Binding Sites , CHO Cells , Catalytic Domain , Cell Membrane/metabolism , Cricetinae , Dose-Response Relationship, Drug , Electrophoresis, Polyacrylamide Gel , Enzyme Precursors/chemistry , Factor Xa/chemistry , Humans , Hydrolysis , Kinetics , Models, Biological , Models, Chemical , Mutation , Phospholipids/metabolism , Protein Binding , Protein Structure, Tertiary , Surface Plasmon Resonance , Ticks , Time Factors
2.
Bioorg Med Chem Lett ; 10(8): 745-9, 2000 Apr 17.
Article in English | MEDLINE | ID: mdl-10782677

ABSTRACT

A novel series of rigid P3-guanylpiperidine peptide mimics 3-14 was designed as potential factor Xa and prothrombinase inhibitors. Incorporation into a P2-gly-P1-argininal motif led to highly potent and selective inhibitors. The synthesis and biological activities of these derivatives are reported herein.


Subject(s)
Factor Xa Inhibitors , Guanine/pharmacology , Peptides/chemistry , Piperidines/pharmacology , Serine Proteinase Inhibitors/pharmacology , Cations , Guanine/analogs & derivatives , Guanine/chemistry , Guanine/pharmacokinetics , Molecular Mimicry , Piperidines/chemistry , Piperidines/pharmacokinetics , Serine Proteinase Inhibitors/chemistry , Serine Proteinase Inhibitors/pharmacokinetics
3.
Bioorg Med Chem Lett ; 9(17): 2573-8, 1999 Sep 06.
Article in English | MEDLINE | ID: mdl-10498211

ABSTRACT

Rigid benzolactam P3-P2 dipeptide mimics were designed and prepared as potential inhibitors of blood coagulation factor Xa. Methoxy substitution of the tetrahydrobenzazepinone scaffold led to potent and selective inhibitors. The synthesis and biological activities of these derivatives are reported herein.


Subject(s)
Factor Xa Inhibitors , Lactams/pharmacology , Serine Proteinase Inhibitors/pharmacology , Drug Design , Lactams/chemical synthesis , Lactams/chemistry , Serine Proteinase Inhibitors/chemical synthesis , Serine Proteinase Inhibitors/chemistry
4.
Biochemistry ; 36(1): 181-91, 1997 Jan 07.
Article in English | MEDLINE | ID: mdl-8993332

ABSTRACT

The prothrombinase complex assembles through reversible interactions between the protease, factor Xa, the cofactor, factor Va, and acidic phospholipid membranes in the presence of calcium ions. Changes in macromolecular recognition by factor Xa which may result from its interaction with factor Va in the prothrombinase complex have been probed using a recombinant derivative of tick anticoagulant peptide where Arg3 has been replaced with Ala (R3A-TAP). In contrast to the wild type inhibitor, R3A-TAP was a weak competitive inhibitor of factor Xa (Ki = 794 nM). The inhibition of the prothrombinase complex by R3A-TAP was characterized by slow, tight-binding kinetics with an increased affinity of approximately 4000-fold (Ki* = 0.195 nM) relative to that of solution-phase factor Xa. Stopped-flow measurements using p-aminobenzamidine (PAB) demonstrated that the reaction between solution-phase factor Xa and R3A-TAP could be adequately described by a single reversible step with rate constants that were consistent with equilibrium binding measurements. The rate-limiting bimolecular combination of R3A-TAP and factor Xa was competitive with PAB binding of the protease. In contrast, the reaction of R3A-TAP with prothrombinase measured using PAB yielded biphasic stopped-flow traces, indicating a multistep pathway for the reaction of the inhibitor with the enzyme complex. The kinetic measurements were consistent with the initial formation of a ternary complex between R3A-TAP, prothrombinase, and PAB followed by two unimolecular steps which lead to PAB dissociation from the enzyme. In this case, prior occupation of the active site by PAB had no effect on the bimolecular reaction between R3A-TAP and prothrombinase. Thus, the interaction of factor Xa with factor Va on the membrane surface alters recognition of R3A-TAP by the protease, leading to changes in the thermodynamics as well as in the observed kinetic mechanism for the reaction. Therefore, a single amino acid substitution in TAP reveals large changes in macromolecular recognition by factor Xa as a consequence of its interaction with the cofactor within the prothrombinase complex.


Subject(s)
Factor V/antagonists & inhibitors , Factor Va/pharmacology , Factor X/antagonists & inhibitors , Factor Xa/metabolism , Peptides/pharmacology , Animals , Arthropod Proteins , Benzamidines/pharmacology , Binding Sites/genetics , Cattle , Factor V/metabolism , Factor X/metabolism , Intercellular Signaling Peptides and Proteins , Kinetics , Models, Chemical , Mutagenesis, Site-Directed/genetics , Peptides/genetics , Peptides/metabolism , Phosphatidylcholines/metabolism , Protein Binding , Protein Conformation , Serine Proteinase Inhibitors/pharmacology , Spectrometry, Fluorescence
5.
Am J Trop Med Hyg ; 54(5): 475-80, 1996 May.
Article in English | MEDLINE | ID: mdl-8644901

ABSTRACT

A potent and specific inhibitor of the human coagulation protease thrombin was identified in salivary gland extracts of the tsetse fly, Glossina morsitans morsitans, an important vector of African trypanosomiasis. This low molecular weight peptide (MW = 3,530 Da as determined by laser desorption mass spectrometry) was purified using a combination of size-exclusion chromatography and reverse-phase, high-performance liquid chromatography, respectively. Amino terminal sequencing of the purified protein reveals no homology to any previously identified serine protease inhibitor or naturally occurring anticoagulant. The tsetse thrombin inhibitor (TTI) is a stoichiometric inhibitor of thrombin, with an apparent equilibrium dissociation inhibitory constant (Ki*) [corrected] of 584 x 10(-15)M. In addition, it is also a potent inhibitor of thrombin-induced platelet aggregation. Like other hematophagous arthropods, tsetse flies appear to have evolved a novel protease inhibitor capable of antagonizing host hemostasis and facilitating blood feeding.


Subject(s)
Antithrombin III/isolation & purification , Saliva/chemistry , Serine Proteinase Inhibitors/isolation & purification , Tsetse Flies , Amino Acid Sequence , Animals , Antithrombin III/chemistry , Antithrombin III/physiology , Female , Humans , Male , Molecular Sequence Data , Molecular Weight , Partial Thromboplastin Time , Platelet Aggregation , Serine Proteinase Inhibitors/chemistry , Serine Proteinase Inhibitors/physiology , Substrate Specificity
6.
Proc Natl Acad Sci U S A ; 93(5): 2149-54, 1996 Mar 05.
Article in English | MEDLINE | ID: mdl-8700900

ABSTRACT

Hookworms are hematophagous nematodes that infect a wide range of mammalian hosts, including humans. There has been speculation for nearly a century as to the identity of the anticoagulant substances) used by these organisms to subvert host hemostasis. Using molecular cloning, we describe a family of potent small protein (75-84 amino acids) anticoagulants from the hookworm Ancylostoma caninum termed AcAP (A. caninum anticoagulant protein). Two recombinant AcAP members (AcAP5 and AcAP6) directly inhibited the catalytic activity of blood coagulation factor Xa (fXa), while a third form (AcAPc2) predominantly inhibited the catalytic activity of a complex composed of blood coagulation factor VIIa and tissue factor (fVIIa/TF). The inhibition of fVIIa/TF was by a unique mechanism that required the initial formation of a binary complex of the inhibitor with fXa at a site on the enzyme that is distinct from the catalytic center (exo-site). The sequence of AcAPc2 as well as the utilization of an exo-site on fXa distinguishes this inhibitor from the mammalian anticoagulant TFPI (tissue factor pathway inhibitor), which is functionally equivalent with respect to fXa-dependent inhibition of fIIa/TF. The relative sequence positions of the reactive site residues determined for AcAP5 with the homologous regions in AcAP6 and AcAPc2 as well as the pattern of 10 cysteine residues present in each of the inhibitors suggest that the AcAPs are distantly related to the family of small protein serine protease inhibitors found in the nonhematophagous nematode Ascaris lumbricoides var. suum.


Subject(s)
Ancylostoma/enzymology , Blood Coagulation Factors/antagonists & inhibitors , Blood Coagulation , Helminth Proteins/genetics , Serine Proteinase Inhibitors/genetics , Amino Acid Sequence , Ancylostoma/genetics , Animals , Binding Sites , Cloning, Molecular , DNA, Complementary/genetics , Molecular Sequence Data , Sequence Alignment , Thromboplastin/metabolism
7.
Bioorg Med Chem ; 3(8): 1009-17, 1995 Aug.
Article in English | MEDLINE | ID: mdl-7582975

ABSTRACT

The low molecular weight alpha-keto amide inhibitor CVS-1347, benzyl-SO2-Met(O2)-Pro-Arg(CO)((CONH)CH2)-phenyl, is a slow, tight binding inhibitor of alpha-thrombin amidolytic activity having a Ki = 1.28 x 10(-10) M. A complex between human alpha-thrombin and a hydrolysis product of CVS-1347 has been determined and refined using crystallography. The crystals belong to monoclinic space group C2 with cell dimensions of a = 71.08, b = 72.05 and c = 72.98 A and beta = 100.8 degrees. The structure was solved using isomorphous replacement methods and refined with resolution limits of (8.00-1.76) A to an R-value of 0.162. The Pro-Arg core of the inhibitor binds in the S2 and S1 subsites respectively, as is usually observed for Pro-Arg thrombin inhibitors. The Met(O2) side chain does not make any close contacts with the enzyme but influences the conformation of Glu192; the N-terminal benzylsulfonyl group makes an aromatic-aromatic contact with Trp215 in the hydrophobic part of the active site. The alpha-keto carboxylic acid of the proteolyzed inhibitor binds with the carboxylate group in the oxyanion hole, demonstrating that this region can accommodate an anion in a protease-peptide complex. The alpha-keto carbonyl group interacts closely with the two most important residues in the active site: the carbon atom is within a covalent bond distance of the active site Ser195 O gamma and the carbonyl oxygen is hydrogen bonded to His57.(ABSTRACT TRUNCATED AT 250 WORDS)


Subject(s)
Antithrombins/chemistry , Oligopeptides/chemistry , Protein Conformation , Thrombin/antagonists & inhibitors , Thrombin/chemistry , Amino Acid Sequence , Antithrombins/chemical synthesis , Antithrombins/pharmacology , Binding Sites , Crystallography, X-Ray , Fibrinolysin/antagonists & inhibitors , Humans , Indicators and Reagents , Kinetics , Molecular Conformation , Molecular Sequence Data , Molecular Structure , Oligopeptides/chemical synthesis , Oligopeptides/pharmacology
8.
Proc Natl Acad Sci U S A ; 92(13): 6152-6, 1995 Jun 20.
Article in English | MEDLINE | ID: mdl-7597095

ABSTRACT

Human hookworm infection is a major cause of gastrointestinal blood loss and iron deficiency anemia, affecting up to one billion people in the developing world. These soil-transmitted helminths cause blood loss during attachment to the intestinal mucosa by lacerating capillaries and ingesting extravasated blood. We have isolated the major anticoagulant used by adult worms to facilitate feeding and exacerbate intestinal blood loss. This 8.7-kDa peptide, named the Ancylostoma caninum anticoagulant peptide (AcAP), was purified by using a combination of ion-exchange chromatography, gel-filtration chromatography, and reverse-phase HPLC. N-terminal sequencing of AcAP reveals no homology to any previously identified anticoagulant or protease inhibitor. Single-stage chromogenic assays reveal that AcAP is a highly potent and specific inhibitor of human coagulation, with an intrinsic K*i for the inhibition of free factor Xa of 323.5 pM. In plasma-based clotting time assays, AcAP was more effective at prolonging the prothrombin time than both recombinant hirudin and tick anticoagulant peptide. These data suggest that AcAP, a specific inhibitor of factor Xa, is one of the most potent naturally occurring anticoagulants described to date.


Subject(s)
Ancylostoma/physiology , Factor Xa Inhibitors , Helminth Proteins/pharmacology , Serine Proteinase Inhibitors/pharmacology , Amino Acid Sequence , Animals , Arthropod Proteins , Blood Coagulation/drug effects , Chromatography, Gel , Chromatography, High Pressure Liquid , Chromatography, Ion Exchange , Helminth Proteins/chemistry , Helminth Proteins/isolation & purification , Hirudins/pharmacology , Humans , Intercellular Signaling Peptides and Proteins , Kinetics , Mass Spectrometry , Molecular Sequence Data , Peptide Fragments/chemistry , Peptide Fragments/isolation & purification , Peptides/pharmacology , Prothrombin Time , Recombinant Proteins/metabolism , Recombinant Proteins/pharmacology , Ticks
9.
Biochemistry ; 33(25): 7897-907, 1994 Jun 28.
Article in English | MEDLINE | ID: mdl-8011652

ABSTRACT

The interaction of factor Xa with factor Va on a membrane surface results in the assembly of the prothrombinase complex. The highly specific and multistep interaction between recombinant tick anticoagulant peptide (rTAP) and factor Xa was used to probe perturbations in the macromolecular interaction sites of factor Xa that accompany prothrombinase assembly. Steady-state kinetic studies indicated that the incorporation of factor Xa into prothrombinase resulted in a modest 3-fold increase in the rate constant for inhibition by rTAP. However, the overall dissociation constant for the enzyme-inhibitor interaction (Ki*) was decreased approximately 30-fold to 5.3 pM. This finding was verified by fluorescence stopped-flow studies of the multistep reaction between rTAP and solution-phase factor Xa or prothrombinase by using 4-aminobenzamidine. The second-order rate constant for the binding or rTAP to the protease (k + 1 = 3.35 x 10(6) M-1.s-1) was increased approximately 2-fold (k + 1 = 6.6 x 10(6) M-1.s-1) following the assembly of prothrombinase, while the rate constant for the subsequent slow displacement of the fluorophore from the active site of factor Xa was decreased by 20-fold. Therefore, factor Va alters macromolecular interaction sites on factor Xa; which leads to the stabilization of intermediates in the reaction of the protease with rTAP and an increased overall affinity for the inhibition of factor Xa. Fluorescence measurements of prothrombinase assembly using factor Xa modified with dansylglutamylglycinylarginine chloromethyl ketone (DEGR-Xa) indicated that the preformed rTAP-Xa binary complex bound to factor Va more tightly (Kd = 30.7 +/- 6.2 pM) than factor Xa alone (Kd = 1.25 +/- 0.29 nM). The 30-fold higher affinity of the rTAP-Xa complex for factor Va can completely account for the increased affinity of rTAP for prothrombinase and implies adequate thermodynamic description of the reactions involved. Collectively, the data suggest that the interaction of factor Xa with factor Va on a membrane surface alters macromolecular recognition sites on factor Xa involved in binding rTAP. As a result of this conformational change, the inhibition of factor Xa by rTAP is thermodynamically favored when the enzyme is assembled in the prothrombinase complex.


Subject(s)
Factor Xa Inhibitors , Peptides/pharmacology , Thromboplastin/chemistry , Animals , Arthropod Proteins , Binding, Competitive , Cattle , Factor Va/metabolism , In Vitro Techniques , Intercellular Signaling Peptides and Proteins , Kinetics , Macromolecular Substances , Protein Binding , Recombinant Proteins , Thermodynamics
10.
J Biol Chem ; 267(25): 17726-31, 1992 Sep 05.
Article in English | MEDLINE | ID: mdl-1387641

ABSTRACT

The vampire bat salivary plasminogen activator (BatPA) is virtually inactive toward Glu-plasminogen in the absence of a fibrin-like cofactor, unlike human tissue-type plasminogen activator (tPA) (the kcat/Km values were 4 and 470 M-1 s-1, respectively). In the presence of fibrin II, tPA and BatPA activated Glu-plasminogen with comparable catalytic efficiencies (158,000 and 174,000 M-1 s-1, respectively). BatPA's cofactor requirement was partially satisfied by polymeric fibrin I (54,000 M-1 s-1), but monomeric fibrin I was virtually ineffective (970 M-1 s-1). By comparison, a variety of monomeric and polymeric fibrin-like species markedly enhanced tPA-mediated activation of Glu-plasminogen. Fragment X polymer was 2-fold better but 9-fold worse as cofactor for tPA and BatPA, respectively, relative to fibrin II. Fibrinogen, devoid of plasminogen, was a 10-fold better cofactor for tPA than fibrinogen rigorously depleted of plasminogen, Factor XIII, and fibronectin; the enhanced stimulatory effect of the less-purified fibrinogen was apparently due to the presence of Factor XIII. By contrast, the two fibrinogen preparations were equally poor cofactors of BatPA-mediated activation of Glu-plasminogen. BatPA possessed only 23 and 4% of the catalytic efficiencies of tPA and two-chain tPA, respectively, in hydrolyzing the chromogenic substrate Spectrozyme tPA. However in the presence of fibrin II, BatPA and tPA exhibited similar kcat/Km values for the hydrolysis of Spectrozyme tPA. Our data revealed that BatPA, unlike tPA, displayed a strict and fastidious requirement for polymeric fibrin I or II. Consequently, BatPA may preferentially promote plasmin generation during a narrow temporal window of fibrin formation and dissolution.


Subject(s)
Fibrin/metabolism , Plasminogen Activators/metabolism , Salivary Glands/enzymology , Tissue Plasminogen Activator/metabolism , Animals , Chiroptera , Factor X/metabolism , Fibrinolysin/metabolism , Humans , Kinetics , Recombinant Proteins/metabolism
11.
Blood ; 76(12): 2560-4, 1990 Dec 15.
Article in English | MEDLINE | ID: mdl-2124935

ABSTRACT

The vampire bat salivary plasminogen activator (Bat-PA) is a potent PA that exhibits remarkable selectivity toward fibrin-bound plasminogen (Gardell et al, J Biol Chem 256: 3568, 1989). Herein, we describe the activity of recombinant DNA-derived Bat-PA (rBat-PA) in a human plasma milieu. rBat-PA and recombinant human single-chain tissue plasminogen activator (rt-PA) are similarly efficacious at lysing plasma clots. In stark contrast to rt-PA, the addition of 250 nmol/L rBat-PA to plasma in the absence of a clot failed to deplete plasminogen, alpha 2-antiplasmin and fibrinogen. The lytic activities exhibited by finger-domain minus Bat-PA (F- rBat-PA) and finger and epidermal growth factor-like domains minus Bat-PA (FG- rBat-PA) were less than rBat-PA, especially at low concentrations of PA; nevertheless, these truncated forms also possessed a strict requirement for a fibrin cofactor. The loss of PA activity following the addition of rBat-PA to plasma was slower than that observed when either rt-PA or two-chain rt-PA was added. The efficacy, fibrin selectivity, and decreased susceptibility to inactivation exhibited by rBat-PA in vitro in a human plasma milieu suggests that rBat-PA may be superior to rt-PA for the treatment of thrombotic complications.


Subject(s)
Chiroptera/physiology , Fibrin/metabolism , Plasma/metabolism , Plasminogen Activators/metabolism , Saliva/chemistry , Tissue Plasminogen Activator/metabolism , Animals , Fibrin/analysis , Fibrinogen/metabolism , Humans , Plasma/chemistry , Plasminogen/metabolism , Plasminogen Activators/analysis , Plasminogen Activators/physiology , Plasminogen Inactivators/pharmacology , Tissue Plasminogen Activator/analysis , Tissue Plasminogen Activator/physiology
12.
Protein Eng ; 2(5): 371-7, 1989 Jan.
Article in English | MEDLINE | ID: mdl-2494653

ABSTRACT

We have designed and constructed a DNA sequence encoding human tissue plasminogen activator (tPA) with convenient restriction sites that flank each of the domains of the heavy chain. To accomplish this, the first 1095 bases of the gene coding for the mature protein were synthesized with unique restriction sites engineered into the interdomainal regions. This synthetic construction was then ligated to a cDNA fragment of the tPA gene that encoded the active site, thus generating a full-length tPA gene. The gene products produced by Chinese hamster ovary (CHO) cells transfected with either the tPA cassette gene or the tPA cDNA gene were then compared with the tPA produced by Bowes melanoma cells to determine whether or not synthetic interdomainal amino acid changes had an effect on the biochemical characteristics of the molecule. Specifically, molecular weight, specific activity, enhancement by fibrinogen fragments and kinetic constants were analysed. None of the properties examined were significantly different from those of the native melanoma tPA. Therefore, the cassette gene described herein should provide considerable versatility and precision in the construction of tPA mutants by facilitating the manipulation of the finger, growth factor and kringle domains, and likewise should be useful in assessing the function of these domains within the tPA molecule. We present this cassette gene system as a model for the analysis of protein domain function applicable to other multi-domain proteins.


Subject(s)
Genes, Synthetic , Proteins , Tissue Plasminogen Activator/genetics , Amino Acid Sequence , DNA Replication , Hydrolysis , Kinetics , Molecular Sequence Data
13.
Enzyme ; 40(2-3): 122-9, 1988.
Article in English | MEDLINE | ID: mdl-3139401

ABSTRACT

Deletion mutants of human tissue plasminogen activator (tPA) were expressed in Chinese hamster ovary cells. These cells had been transfected with genes that encoded tPA but included restriction sites that allowed the deletion of DNA encoding specific structural domains of the tPA molecule's heavy chain. Purified, two-chain mutant tPAs, or analogues of tPA, lacking one or several structural domains, along with Bowes melanoma tPA were studied in order to determine their susceptibility to inhibition by plasminogen activator inhibitor-1 (PAI-1). The full-length analogue of tPA, designated by its domains FGK1K2P, as well as analogues GK1K2P, FK2P, and FGK1P were treated with various amounts of PAI-1. When the amounts of added tPA and analogues were standardized so that each generated the same absorbance in a chromogenic assay containing S-2251, plasminogen, and fibrinogen fragments, there was a significant difference in the way in which the analogues were titrated by the inhibitor. The melanoma tPA and FGK1K2P were the most susceptible, FK2P slightly less sensitive, and GK1K2P and FGK1P the least sensitive to inhibition. In contrast, when the amounts of enzyme used were standardized on the basis of absorbance generated in a direct assay employing the chromogenic substrate S-2288 and then titrated with PAI-1, these differences in susceptibility to inhibition were not observed. Based on these data, the differential susceptibility to inhibition observed in the plasminogen-dependent assay was attributed to the extent to which the activity of a given analogue is enhanced by fibrinogen fragments, and thus reflected the different amounts of enzymes added in order to standardize the assay on the basis of absorbance.(ABSTRACT TRUNCATED AT 250 WORDS)


Subject(s)
Tissue Plasminogen Activator/genetics , Chromosome Deletion , Fibrinogen/metabolism , Glycoproteins/pharmacology , Humans , Peptide Fragments/metabolism , Plasminogen Inactivators , Recombinant Proteins/pharmacology , Tissue Plasminogen Activator/analogs & derivatives
14.
Br J Haematol ; 64(2): 283-90, 1986 Oct.
Article in English | MEDLINE | ID: mdl-2946315

ABSTRACT

Plasma levels of protein C and protein S antigens were measured in eight children who developed thrombosis following asparaginase-prednisone-vincristine treatment for acute lymphoblastic leukaemia and in nine similarly treated children without this complication. Protein C antigen levels were below normal in three of the eight patients with thrombosis and in three of the nine patients without the complication (P = 0.38). Low protein S antigen levels were found in five of six patients with thrombosis and in two of seven patients without thrombosis (P = 0.10). Plasma factor IX and factor X antigen levels, other vitamin K dependent factors, were also measured in the two groups of patients. In general, reduced levels of protein C, protein S or both antigens (anticoagulant vitamin K dependent proteins) were associated with reduced levels of factor IX, factor X, or both of these factors (procoagulant vitamin K dependent clotting proteins). The ratios of protein C and protein S antigens to each other and to factor IX and factor X antigens did not differ between the two groups. Thus, there is no clear evidence that reduced levels of protein C and (or) protein S cause thrombosis in leukaemia patients treated with this drug combination.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/adverse effects , Glycoproteins/metabolism , Leukemia, Lymphoid/drug therapy , Protein C/metabolism , Thrombosis/chemically induced , Adolescent , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Asparaginase/administration & dosage , Asparaginase/adverse effects , Child , Child, Preschool , Factor IX/metabolism , Factor X/metabolism , Female , Humans , Leukemia, Lymphoid/blood , Male , Prednisone/administration & dosage , Prednisone/adverse effects , Protein S , Thrombosis/blood , Vincristine/administration & dosage , Vincristine/adverse effects
15.
Am J Clin Pathol ; 82(5): 573-81, 1984 Nov.
Article in English | MEDLINE | ID: mdl-6548600

ABSTRACT

Specific radioimmunoassays, sensitive to plasma levels of less than 1% of normal, were developed for protein C and Factor X. In 31 normal subjects, mean plasma antigen levels were as follows: protein C, 3.23 +/- 0.79 microgram/mL (2 SD); Factor X, 7.74 +/- 1.81 microgram/mL. In patient son chronic warfarin therapy, protein C and factor X were depressed equivalently: protein C, 42% +/- 20% (of a pooled plasma reference); Factor X, 44% +/- 24%. Protein C antigen fell much more rapidly than Factor X antigen when warfarin therapy was begun, creating an initial period of potential hypercoagulability. In patients with severe liver disease, mean protein C antigen (25% +/- 17%) was lower than Factor X antigen (51% +/- 29%). Protein C antigen levels did not appear to be a sensitive indicator of compensated intravascular coagulation or systemic fibrinolysis induced by infusion of streptokinase. Clinical implications of these findings are discussed.


Subject(s)
Blood Coagulation Factors/analysis , Factor X/analysis , Glycoproteins/analysis , Hematologic Diseases/immunology , Cross Reactions , Disseminated Intravascular Coagulation/immunology , Female , Hemostasis , Humans , Kinetics , Liver Diseases/immunology , Lupus Erythematosus, Systemic/immunology , Pregnancy , Protein C , Radioimmunoassay , Streptokinase/pharmacology , Warfarin/pharmacology
16.
J Immunol Methods ; 51(3): 293-305, 1982.
Article in English | MEDLINE | ID: mdl-6809833

ABSTRACT

We report here the successful labeling of rabbit peripheral blood neutrophils with [111In]oxine. We found that standard techniques for preparation of rabbit neutrophils, while acceptable for maintenance of in vitro function, rendered the neutrophils ineffective for in vivo use after labeling with 111In. Specifically, rabbit neutrophils were sensitive to the use of hypotonic shock for red cell elimination, centrifugation into a button during preparation, and the presence of oxine during chemotaxis in vitro. Using a carefully modified method of neutrophil preparation and labeling, we found that 111In-labeled rabbit neutrophils retained normal in vitro function, including chemotaxis. In addition, using our method, 34% +/- 5% of labeled neutrophils were recoverable in peripheral blood 5 min after intravenous injection. The half-life of circulating radio-labeled neutrophils was 5.6 +/- 2 h. Continuous external imaging of radio-labeled neutrophils after intravenous injection showed initial lung uptake, followed by rapid clearance of radioactivity in the lungs (50% clearance in 10.5 +/- 3.3 min.) Hepatic radioactivity was maximal by 30 min after injection and thereafter slowly declined. Finally, we found that 111In-labeled rabbit neutrophils migrated to sites of artificially induced inflammation. Our findings indicate that 111In-labeled rabbit neutrophils, if prepared under optimal conditions, should provide a useful tool for investigating the fate of neutrophils in experimental inflammatory conditions in this animal.


Subject(s)
Hydroxyquinolines , Indium , Neutrophils/metabolism , Oxyquinoline , Radioisotopes , Animals , Cell Survival , Chemotaxis, Leukocyte/drug effects , Female , Inflammation/diagnosis , Male , N-Formylmethionine/analogs & derivatives , N-Formylmethionine/pharmacology , N-Formylmethionine Leucyl-Phenylalanine , Oligopeptides/pharmacology , Oxyquinoline/pharmacology , Rabbits , Skin/metabolism , Sodium Chloride/pharmacology , Time Factors , Zymosan/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL